Lymphomas: Promotes proliferation in Hodgkin’s/Reed-Sternberg cells while inducing apoptosis in select non-Hodgkin’s lines
Viral Infections: TNFSF8 polymorphisms (rs3181366-T, rs2295800-C) correlate with HCV susceptibility
Cardiovascular: rs927374/rs2295800 linked to 16% reduction in neutrophil counts post-myocardial infarction
Polymorphism | Association | Population Studied | P-value |
---|---|---|---|
rs927374 (GG) | ↓ Neutrophil count | Post-MI patients | 5.1×10⁻⁵ |
rs3181366 (T) | ↑ HCV risk | Chinese cohort | 0.002 |
rs2295800 (C) | ↑ HCV risk | High-risk groups | 0.012 |
Antibody Clone | Target Region | Staining Application |
---|---|---|
MAB10281 | Extracellular | IHC (spleen, granulocytes) |
AF1028 | Soluble domain | Flow cytometry (PBMCs) |
TNFSF8, also known as CD30 Ligand, is a member of the tumor necrosis factor superfamily that plays significant roles in immune regulation. It functions primarily through binding to CD30 receptor (TNFRSF8), triggering signaling cascades that influence cell survival, proliferation, and cytokine production . In experimental studies, TNFSF8 has been shown to induce IL-6 secretion in human Hodgkin's lymphoma cell lines, demonstrating its role in inflammatory responses .
Methodologically, researchers studying TNFSF8 function typically employ flow cytometry for detection in human PBMCs (peripheral blood mononuclear cells), especially after stimulation with agents like PMA and Calcium Ionomycin . Functional studies often utilize neutralizing antibodies to block TNFSF8 activity in controlled cellular environments.
TNFSF8 expression can be detected through multiple methodological approaches:
Flow cytometry: Human PBMCs can be treated with PMA (50 ng/mL) and Calcium Ionomycin (200 ng/mL) overnight to induce expression, then stained with specific anti-TNFSF8 monoclonal antibodies .
mRNA expression analysis: Transcriptomic approaches like RNA-seq or microarray analysis are commonly used in larger studies, as evidenced by TCGA breast cancer data analysis .
Functional assays: TNFSF8 activity can be measured through its ability to induce IL-6 secretion in specific cell lines like HDLM human Hodgkin's lymphoma cells .
For reliable detection, cross-validation with multiple methods is recommended, particularly when studying different tissue types or disease states.
Research has identified significant associations between specific TNFSF8 polymorphisms and neutrophil counts in post-myocardial infarction (MI) patients. The polymorphisms rs927374 (P=5.1 x 10⁻⁵) and rs2295800 (P=1.3 x 10⁻⁴) show strong associations with neutrophil counts . These single-nucleotide polymorphisms (SNPs) are in high linkage disequilibrium (r²=0.97), suggesting they may represent the same functional effect .
For rs927374, the data shows a clear genotype-dependent effect on neutrophil counts:
Genotype | Mean Neutrophil Count (±SD) | Percent Difference from CC |
---|---|---|
CC | 9.0 ± 5.2 | Reference |
GG | 7.6 ± 5.1 | 16% lower |
This association persisted after controlling for various clinical characteristics and remained unchanged after adjusting for case-control status . These findings suggest that genetic variation in TNFSF8 may influence the post-MI inflammatory response, offering potential insights into personalized cardiovascular disease management.
Methodologically, researchers investigating such associations should consider:
Genotyping multiple SNPs in the TNFSF8 region
Controlling for relevant clinical variables
Performing linkage disequilibrium analysis to identify functional variants
Validating findings in independent cohorts
When analyzed by breast cancer subtype, TNFSF8 expression shows a significant correlation with OS in multiple subtypes:
Luminal A: Higher expression correlates with better OS
Luminal B: Higher expression correlates with better OS
Basal-Like: Higher expression correlates with better OS
These findings suggest TNFSF8 may serve as a prognostic biomarker across different breast cancer subtypes .
For methodology in survival analysis studies:
Use Kaplan-Meier analysis with appropriate statistical testing
Stratify by molecular subtypes (Luminal A, Luminal B, HER2+, Basal-Like)
Validate findings using independent cohorts
Consider multivariate analysis to account for clinical covariates
Analysis using the TIMER database indicates that TNFSF8 expression positively correlates with infiltration of multiple immune cell types in breast cancer, including B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils, and dendritic cells (all with P<0.05) . This finding suggests TNFSF8 may play a role in modulating the tumor immune microenvironment.
Researchers investigating this relationship should:
Use computational deconvolution methods to estimate immune cell infiltration from bulk RNA-seq data
Validate computational findings with immunohistochemistry or flow cytometry
Perform correlation analyses between gene expression and immune cell markers
Consider single-cell RNA-seq to further resolve immune cell populations and their relationship with TNFSF8
Several methodological approaches have proven effective for studying TNFSF8 function:
Neutralization assays: The standard approach involves measuring IL-6 secretion induced by recombinant human TNFSF8 in HDLM human Hodgkin's lymphoma cells, using a cross-linking antibody (e.g., Mouse polyHistidine Monoclonal Antibody at 10 μg/mL) . Neutralization is then assessed by adding increasing concentrations of anti-TNFSF8 antibody.
Flow cytometry: For detecting TNFSF8 expression on cell surfaces, researchers typically use anti-TNFSF8 monoclonal antibodies on stimulated cells (often using PMA and Calcium Ionomycin) .
Gene expression manipulation: Knockdown or overexpression studies using siRNA, CRISPR-Cas9, or overexpression vectors can help determine TNFSF8's functional impact.
The optimal concentration for neutralization experiments (ND₅₀) is typically 1-4 μg/mL of anti-TNFSF8 antibody in the presence of 1 μg/mL recombinant human TNFSF8 . Researchers should always include appropriate controls, including isotype controls for antibody experiments.
When investigating TNFSF8 genetic variants in population studies, researchers should consider:
SNP selection strategy:
Sample size considerations:
Statistical analysis approach:
Control for population stratification using principal component analysis
Adjust for relevant clinical covariates
Consider multiple testing correction (e.g., Bonferroni or False Discovery Rate)
Validate findings in independent cohorts
Functional validation:
Follow-up significant associations with in vitro studies
Use reporter assays to assess regulatory effects of variants
Consider eQTL analysis to link variants to expression differences
When faced with contradictory findings regarding TNFSF8 across different cancer studies, researchers should consider several methodological factors:
Cancer type specificity: TNFSF8's role may differ fundamentally between cancer types. In breast cancer, high expression correlates with better survival outcomes , but this pattern may not hold for other malignancies.
Subtype heterogeneity: Even within a single cancer type, molecular subtypes can show different relationships with TNFSF8. Analysis of breast cancer subtypes shows that higher TNFSF8 expression correlates with better OS in Luminal A, Luminal B, and Basal-Like subtypes .
Analysis methodology differences: Varying statistical approaches, cutoff definitions for "high" versus "low" expression, and adjustment for covariates can lead to apparently contradictory results.
Context-dependent function: TNFSF8 may have different or even opposing effects depending on the immune microenvironment, which itself varies across cancer types and stages.
To reconcile contradictions, researchers should:
Perform meta-analyses with careful attention to methodological differences
Stratify analyses by cancer type, subtype, and stage
Consider interaction effects with other immune markers
Validate findings using multiple independent datasets
The positive correlation between TNFSF8 expression and immune cell infiltration in breast cancer has several important implications:
Researchers should note that correlation does not imply causation, and functional studies are needed to determine whether TNFSF8 directly influences immune cell recruitment or whether its expression is simply a marker of an immunologically "hot" tumor microenvironment.
For optimal detection of TNFSF8 protein in clinical samples, researchers should consider:
Sample preparation:
Detection methods:
Controls and validation:
Quantification approaches:
For flow cytometry: Report mean fluorescence intensity ratios
For IHC: Use standardized scoring systems (H-score, Allred score)
Consider digital pathology for more objective quantification
To investigate the functional consequences of TNFSF8 polymorphisms such as rs927374 and rs2295800 , researchers should employ a multi-faceted approach:
Expression quantitative trait loci (eQTL) analysis:
Assess whether the polymorphisms affect TNFSF8 mRNA or protein expression levels
Use available databases (GTEx, eQTLGen) to explore existing associations
Perform tissue-specific eQTL analysis when possible
In vitro functional assays:
Protein function studies:
Assess whether variants affect protein-protein interactions
Measure downstream signaling pathway activation
Evaluate cellular responses like cytokine production or proliferation
In silico prediction:
Use computational tools to predict functional effects of coding variants
Analyze potential effects on transcription factor binding for regulatory variants
Evaluate evolutionary conservation at variant positions
Based on current knowledge, several promising research directions for TNFSF8 include:
Precision medicine applications:
Mechanistic studies:
Clinical correlations:
Expanding association studies to diverse populations
Investigating TNFSF8's role in additional disease states
Determining whether TNFSF8 can predict response to immunotherapies
Technical innovations:
Developing improved detection methods for TNFSF8 protein and genetic variants
Creating animal models with human-relevant TNFSF8 mutations
Exploring single-cell approaches to better understand cell type-specific effects
CD30 Ligand is a type II membrane protein, meaning it has a single transmembrane domain with its N-terminus located inside the cell and its C-terminus outside . The human recombinant form of CD30 Ligand is typically produced in a laboratory setting using various expression systems, such as HEK293 cells or mouse myeloma cell lines . The recombinant protein is often tagged with a histidine tag to facilitate purification and detection .
CD30 Ligand interacts specifically with its receptor, CD30, which is a member of the TNF receptor superfamily . This interaction plays a crucial role in the regulation of immune responses, including cell proliferation, activation, differentiation, and apoptosis . CD30-CD30 Ligand signaling is particularly important in the context of T cell-dependent immune responses and has been implicated in various pathological conditions, including Hodgkin lymphoma, large cell anaplastic lymphomas, and Burkitt lymphomas .
Due to its significant role in immune regulation and its involvement in various hematologic malignancies, CD30 Ligand and its receptor CD30 are considered potential therapeutic targets . Research into CD30 Ligand has led to the development of therapeutic antibodies and other biologics aimed at modulating CD30-CD30 Ligand interactions for the treatment of cancers and autoimmune diseases .
In the laboratory, recombinant CD30 Ligand is used in various assays to study its biological activities. For example, it has been shown to stimulate interleukin-6 (IL-6) secretion by Hodgkin’s lymphoma cells . This makes it a valuable tool for understanding the mechanisms underlying immune responses and for developing new therapeutic strategies.